MCB Accepts, published online ahead of print on 29 September 2014 Mol. Cell. Biol. doi:10.1128/MCB.00524-14 Copyright © 2014, American Society for Microbiology. All Rights Reserved.

1

Paip1, an effective stimulator of translation initation, is targeted by

2

WWP2 for ubiquitination and degradation

3 4

Yanrong Lv1, Kai Zhang1, Haidong Gao1*

5 6

Department of Breast Surgery, QiLu Hospital of Shandong University, Jinan, Shandong,

7

China1

8 9 10

Running title: WWP2 ubiquitylates and degrades Paip 1

11 12 13 14 15

* Corresponding author. Haidong Gao, Department of Breast Surgery, QiLu Hospital of Shandong University No. 107, Wen hua Xi Road, Jinan, China, Tel: +86 15806661668, E-mail: [email protected] 1

16

ABSTRACT

17

Poly(A)-binding protein-interacting protein 1 (Paip1) stimulates translational initiation by

18

inducing the circularization of mRNA. However, the mechanisms underlying Paip1

19

regulation, particularly its protein stability, are still unclear. Here we show that the E6AP

20

carboxyl terminus (HECT)-type ubiquitin ligase WW domain-containing protein 2

21

(WWP2), a homolog of HECT-type ubiquitin ligase WWP1, interacts with and targets

22

Paip1 for ubiquitination and proteasomal degradation. Mapping of the region, including

23

the WW domain of WWP2, revealed the interaction between WWP2 and the

24

PABP-binding motif 2 (PAM2) of Paip1. The two consecutive PxxY motifs in PAM2 are

25

required for WWP2-mediated ubiquitination and degradation. Furthermore, ectopic

26

expression of WWP2 decreases translational stimulatory activity with the degradation of

27

Paip1. We therefore provide the evidence that the stability of Paip1 can be regulated by

28

ubiquitin-mediated degradation; thus, highlighting the importance of WWP2 as a

29

suppressor of translation.

30 31

INTRODUCTION

32

Regulation of gene expression occurs in eukaryotes during messenger RNA (mRNA)

33

translation, specifically at the initiation of translation. Deregulation at this step of the

34

translation process leads to abnormal gene expression, which in turn alters cell growth

35

and possibly leads to cancer development (1–3). Translational initiation comprises a

36

series of discrete steps and starts with the dissociation of 80S ribosomes into subunits. 2

37

The eukaryotic translational initiation factors (eIFs) 1, 1A, and 5, and the eIF3 complex

38

promote the binding of the eIF2-GTP-Met-tRNAi ternary complex to the 40S subunit,

39

thereby forming a 43S pre-initiation complex (PIC) (4–7). The 43S PIC is loaded onto the

40

mRNA near the 5’-7-methylguanosine cap by numerous factors, including eIF3,

41

poly(A)-binding protein (PABP), eIF4B, and eIF4F complex. The eIF4F complex

42

comprises three subunits, namely, a cap-binding protein (eIF4E), an RNA helicase

43

(eIF4A), and a scaffold protein eIF4G (8,9). eIF4G harbors the binding domains for

44

PABP, eIF4E, eIF4A, and eIF3 in mammals. The binding domains for eIF4E and PABP

45

in eIF4G enable the assembly of a stable, circular messenger ribonucleoprotein (mRNP)

46

by eIF4G; eIF4G–eIF3 interaction generates a protein bridge between the mRNPs

47

(10–13).

48

PABP-interacting protein 1 (Paip1) is a PABP-binding protein that contains two

49

distinct PABP-binding motifs (PAMs). PAM1 binds to RNA recognition motif 2 in the N

50

terminus of PABP; PAM2, which is a conserved region comprising approximately 15

51

amino acids, binds to the PABC domain of PABP (14, 15). Paip1 shows 39% similarity

52

to eIF4G and the eIF4G-related protein, p97/DAP5/NAT1 (16–18). A specific portion

53

that is present in both Paip1 and eIF4G has one of two known eIF4A binding regions and

54

an eIF3 binding site (19, 20). The abovementioned findings indicate that Paip1

55

co-immunoprecipitates with eIF4A and eIF3 (21, 22). The presence of Paip1 in animal

56

cells may indicate the involvement of a mechanism that links PABP to eIF4A, thereby

57

causing the circularization of mRNA (21). Data from previous studies suggest that the 3

58

interaction of Paip1 with eIF3 stabilizes circular mRNP conformation, which is formed

59

by eIF4G–PABP interaction (22). eIF3 is reportedly phosphorylated by S6K1/2, which

60

stimulates the Paip1–eIF3 interaction and the initiation of translation (23). Paip1 is an

61

important positive effector of translational initiation, but information is lacking on the

62

mechanism underlying Paip1 regulation.

63

In the present study, we demonstrated the critical function of an E6AP carboxyl

64

terminus (HECT)-domain comprising an E3 ubiquitin ligase WW domain-containing

65

protein 2 (WWP2), which is also known as atrophin-1 interacting protein 2 (AIP2), in the

66

regulation of Paip1 protein stability. WWP2 is homologous to the HECT domain-type

67

ubiquitin-protein ligase and participates in the regulation of craniofacial development and

68

chondrogenesis (24, 25). WWP2 also participates in the maintenance of key oncogenic

69

signaling pathways, which are linked to cancer cell growth, survival, and tumor spread

70

(26, 27). Here we showed that WWP2 interacted with the PAM2 motif of Paip1 via the

71

WW domain. Further investigation revealed that WWP2 targeted Paip1 for ubiquitination

72

and degradation via the “PEFYPSGY” sequence in the PAM2 motif. Importantly, WWP2

73

was found to participate in translational initiation by regulating Paip1 protein level.

74

MATERIALS AND METHODS

75

Cell Culture and Transfection. HEK293T and HeLa cells were cultured in Dulbecco’s

76

modified Eagle’s medium supplemented with 10% fetal bovine serum and 5 U/ml

77

penicillin-streptomycin (Gibco, USA) in 5% CO2. Cells were transfected by

78

Lipofectamine 2000 following the manufacturer’s protocol (Invitrogen, USA). 4

79

Antibodies and Reagents. The proteasomal inhibitors MG132 and lactacystin were

80

purchased from Sigma-Aldrich, USA. The WWP2 and Paip1 antibodies were purchased

81

from Abcam, UK. Anti-glyceraldehyde 3-phosphate dehydrogenase (Anti-GAPDH) and

82

secondary antibodies were purchased from Santa Cruz Biotechnology, Inc., USA.

83

Anti-hemagglutinin (Anti-HA) was obtained from Roche Applied Science, Germany, and

84

anti-Myc and anti-Flag antibodies were from MBL.

85

Immunoprecipitation and Immunoblotting. For general cell lysis, transfected cells

86

were harvested and lysed in HEPES lysis buffer (containing 20 mM HEPES, pH 7.2,

87

50 mM NaCl, 0.5% Triton X-100, 1 mM NaF, and 1 mM dithiothreitol) and boiled with

88

2× SDS–PAGE loading buffer. For immunoprecipitation, cell lysates were prepared in

89

500 ml HEPES buffer supplemented with a protease inhibitor mixture (Roche Applied

90

Science). Immunoprecipitation was performed by primary antibody incubation for 3 h

91

followed by overnight incubation with Protein A/G Sepharose beads (Santa Cruz). The

92

beads were washed with HEPES buffer thrice and examined by immunoblotting.

93

Immunofluorescence. For subcellular localization analyses, cells were fixed with 4%

94

paraformaldehyde and permeabilized in 0.2% Triton X-100 (phosphate-buffered saline).

95

Proteins were stained using the indicated antibodies and detected with a TRITC- or

96

FITC-conjugated secondary antibody. The nuclei were stained with 4′,6′

97

-diamidino-2-phenylindole hydrochloride (DAPI; Sigma), and images were visualized

98

with a Zeiss LSM 510 Meta inverted confocal microscope.

99

RNA Interference. The WWP2 siRNA-1 (5’- CACCTACTTTCGCTTTATA5

100

3’), siRNA-2 (5’- GGAGTACGTGCGCAACTAT-3’), and non-targeting siRNAs

101

(5’-UUCUCCGAACGUGUCACGU-3’) were synthesized by Shanghai GenePharm. All

102

siRNAs were transfected into the cells according to the manufacturer’s protocol.

103

Determination of turnover of Paip1. HEK293T cells were transfected with Myc-Paip1

104

with Flag-WWP2, Flag-WWP2-C838A, or empty vector (or interfering RNA). The cells

105

were treated with cycloheximide (CHX, 15 mg/ml, Sigma) at 36 h after transfection and

106

harvested at the indicated time. The protein level was measured by Western blot using the

107

indicated antibodies.

108

Ubiquitination assays. Cells were transfected with Myc-Paip1 and Flag-WWP2 or its

109

mutants to investigate Paip1 ubiquitination. Subsequently, cells were treated with MG132

110

(20 ìM) for 8 h before harvest. Cell lysis solution was prepared in modified RIPA lysis

111

buffer containing 10 mM Tris–HCl, pH 7.5, 150 mM NaCl, 5 mM

112

ethylenediaminetetraacetic acid (EDTA), 1% NP-40, 1% sodium deoxycholate, 0.025%

113

sodium dodecyl sulfate, and protease inhibitors. Immunoprecipitation was conducted

114

using the indicated antibody. Subsequently, immunoblot analysis was performed. For in

115

vitro ubiquitination assay, His-WWP2 and glutathione s-transferase (GST)-Paip1 were

116

expressed in Escherichia coli and were purified with Ni-nitrilotriacetate-agarose (Qiagen,

117

Netherlands) and glutathione-Sepharose 4B beads (Amersham, UK), respectively. The

118

assays were conducted in 30 il ubiquitination assay buffer (containing 50 mM Tris, pH

119

8.0, 50 mM NaCl, 1 mM dithiothreitol, 5 mM MgCl2, and 3 mM ATP) with 0.7 ìg of E1,

120

1 ìg of UbcH5c (E2), 15 ìg of HA-ubiquitin (Boston Biochem, MA, USA), 0.7 ìg of 6

121

His-WWP2 (wild-type or C838A mutant), and 1.5 ìg GST–Paip1. Samples were

122

incubated at 30 °C for 2 h. Reactions were terminated using the sample buffer.

123

Real-time reverse transcription–polymerase chain reaction (RT–PCR). Total RNA

124

was isolated from the cells using TRIZOL (Invitrogen) and reverse transcribed using 1 ìg

125

of total RNA with an oligo(dT) primer. The following primers were used for real-time

126

PCR: human GAPDH forward,

127

5’-GGGAAGGTGAAGGTCGGAGT-3’; GAPDH reverse, 5’-

128

TTGAGGTCAATGAAGGGGTCA-3’; human WWP2 forward,

129

5’- CGCAACTATGAGCAGTGGCA-3’; human WWP2 reverse,

130

5’- GGTCGTGCGAGTGTTATGGT-3’; human Paip1 forward, 5’-

131

GGAGAACTGGAAAGCCGAGGGTA-3’; and human Paip1 reverse,

132

5’- GTGTAACTGGAAGAATAACCTGAAGGG-3’; Renilla forward

133

5’-CAGTGGTGGGCCAGATGTAAACAA-3’; and Renilla reverse

134

5’-TAATACACCGCGCTACTGGCTCAA-3’.

135

Translation assays. HeLa cells were grown in 60 mm dishes and co-transfected with

136

Myc-Paip1, pTet-HA-WWP2 (WT or CA) or control vector pUHD-15-1 expressing the

137

Tet-controlled transactivator (tTA) (28), and pRL-CMV (Promega, USA). pRL-CMV is a

138

Renilla luciferase reporter construct. The medium was replaced with Tet at a final

139

concentration of 0 or 300 ng/ml after 4 h. Cells were harvested 48 h after transfection,

140

and extracts were used to quantify Renilla luciferase by a dual-luciferase reporter assay

141

system (Promega). Protein concentration was determined using Bio-Rad protein assay 7

142

reagent, and Renilla luciferase activity was corrected according to the protein

143

concentration. The relative induction for each construct was determined by calculating

144

the ratio of Renilla luciferase activity between the induced condition (no Tet) and the

145

repressed condition (with 300 ng/ml Tet). Extracts were subjected to SDS–PAGE and

146

Western blot analysis.

147

Results

148

WWP2 negatively regulates the protein level of Paip1 in a proteasome-dependent

149

manner. To explored the mechanism underlying Paip1 regulation, we first investigated

150

whether or not the Paip1 protein was regulated through the proteasome pathway, which is

151

the most critical pathway in the regulation of cellular protein stability and quality in

152

eukaryotes. The level of endogenous Paip1 protein increased significantly after MG132

153

treatment in both HEK293T and HeLa cells (Fig. 1A), thereby suggesting that Paip1

154

protein regulation depended on the proteasome system. We also examined the effect of

155

MG132 treatment on the levels of other related translation factors, such as PABP and

156

Paip2. The proteasome inhibitors failed to affect the PABP and Paip2 protein levels (Fig.

157

1A). The ubiquitin ligases controlled ubiquitination and determined the specificity of

158

substrate recognition during ubiquitin-mediated proteasomal degradation. So we

159

determined which ubiquitin ligase downregulated Paip1 protein stability. Sequence

160

analysis showed that Paip1 comprised three putative PxxY-type PY motifs (Fig. 1B).

161

These motifs are recognized by the Nedd4 family of ubiquitin ligases. Among the nine

162

members of the Nedd4 ligases, only WWP2 specifically downregulated Paip1 protein 8

163

levels (Fig. 1C). The downregulation of Paip1 by WWP2 was blocked by proteasome

164

inhibitors MG132 and lactacystin (Fig. 1D). Thus, we hypothesized that WWP2 functions

165

as an ubiquitin ligase in Paip1 degradation.

166

To test our hypothesis, we transiently transfected HEK293T cells with a constant

167

amount of Paip1 and an increasing amount of wild-type WWP2 or ligase-inactive

168

WWP2-C838A expression vectors. The Paip1 protein level progressively decreased with

169

increasing WWP2-WT expression, but this phenomenon was not observed with increasing

170

WWP2-C838A mutant expression (Figs. 1E and 1F). No change in the Paip1 mRNA level

171

was observed. Thus, WWP2 could mediate Paip1 destruction depending on its ubiquitin

172

ligase activity.

173

To verify whether endogenous WWP2 regulates Paip1 level, WWP2 was depleted

174

using two independent siRNAs. The depletion of endogenous WWP2 in HEK293T and

175

HeLa cells significantly increased Paip1 protein level (Fig. 1G). However, the mRNA

176

level was not affected by the abovementioned changes (Fig. 1H). WWP2 can function as

177

an ubiquitin ligase to maintain proper Paip1 protein levels in human cells.

178

Paip1 and Paip2 share similar domains and compete for PABP binding. According

179

to previous studies, another HECT-type ubiquitin ligase, namely, EDD, targets Paip2 for

180

degradation during PABP depletion. We examined the effect of WWP2 overexpression

181

on Paip2 and found that WWP2 did not affect Paip2 protein levels (Fig. 1I). Furthermore,

182

overexpression of Paip2 did not affect WWP2-mediated Paip1 degradation (Fig. 1J).

183

Paip1 and Paip2 did not compete for WWP2 binding, and the degradation of the two 9

184

proteins required different ubiquitin ligases. Moreover, the Paip1 binding protein, PABP,

185

was not regulated upon WWP2 silencing and overexpression (Fig. 1I).

186

WWP2 promotes the degradation and ubiquitination of Paip1. To assessed

187

whether the reduction in the protein level of Paip1, which was induced by WWP2

188

overexpression, was due to increasing Paip1 degradation. We analyzed the steady-state

189

levels of Paip1 by applying CHX, an inhibitor of protein synthesis. The half-life of

190

exogenously expressed Paip1 was greatly reduced by the expression of WWP2-WT, but

191

not by the expression of Paip1-C838 A (Fig. 2A). However, the depletion of WWP2

192

prolonged the half-life of endogenous Paip1 (Fig. 2B). Thus WWP2 promoted the

193

degradation of the Paip1 protein.

194

We determined whether WWP2 functioned as an ubiquitin ligase and if WWP2

195

directly promoted the ubiquitination of Paip1. An in vitro ubiquitination assay was

196

initially conducted using purified E1 and UbcH5c, bacteria-expressed His-WWP2-WT or

197

His-WWP2-C838A, and GST-Paip1 or GST (control). Bacteria-expressed and purified

198

WWP2-WT promoted the ubiquitination of Paip1, but such effect on Paip1 was not

199

induced by the WWP2-C838A (Fig. 2C). Overexpressed WWP2 enhanced the

200

ubiquitination of Paip1 in the presence of MG132 in HEK293T cells (Fig. 2D). The

201

required WW and HECT domains were sufficient for WWP2 to promote the degradation

202

of Paip1 (Fig. 2E). These data suggested that WWP2 functioned as a biologically active

203

ubiquitin ligase and induced the ubiquitination and degradation of Paip1.

204

The WW domain of WWP2 is required for binding to the PAM2 motif of Paip1. 10

205

Proteins should at least partly colocalize to be functionally linked. Indirect

206

immunofluorescence assays revealed that WWP2 and Paip1 colocalized predominantly in

207

the cytoplasm (Fig. 3A). We further confirmed the interaction of WWP2 with Paip1 in

208

mammalian cells. A co-immunoprecipitation assay revealed the association between

209

Paip1 and WWP2-WT or C838A mutant (Fig. 3B). Thus, ubiquitin ligase activity was not

210

required for the interaction. In addition, endogenous Paip1 and its binding partners were

211

co-immunoprecipitated with WWP2, but not with the control IgG from HeLa cells

212

(Fig. 3C left panel). Given the interactions between WWP2 and eIF3a, PABP disappeared

213

in a Paip1 depletion cell extract (Fig. 3C right panel), thereby suggesting that eIF3a and

214

PABP bonded to WWP2 depending on the presence of Paip1.

215

The direct association between WWP2 and Paip1 was confirmed by GST pull-down

216

assay. WWP2 protein comprised three domains, namely, C2, WW (including four tandem

217

WW domains), and HECT. The results of the GST pull-down assays indicated that the

218

WW domain mediated the direct interaction between WWP2 and Paip1 (Fig. 3D). The C2

219

or HECT domains of WWP2 did not show the same regulatory action. The

220

abovementioned results contradicted a previous finding, which stated that the WW

221

domains of Nedd4 family members preferred to recruit substrates (29). Paip1 comprised

222

two binding sites for PABP, namely, PAM1 and PAM2 (15). PAM2 was required for the

223

binding of Paip1 to eIF3. Different parts of Paip1 were fused to GST to identify the

224

WWP2-binding site in Paip1. His-WWP2 was detected in reactions involving the

225

full-length protein, the fusion protein with retained Paip1 N terminus (comprising amino 11

226

acids 1 to 143), and the PAM2 motif (comprising amino acids 116 to 143). However, the

227

large C-terminal region (comprising amino acids 144 to 479) was not required for the

228

binding of Paip1 to WWP2 (Fig. 3E). The PAM2 motif was previously identified in

229

numerous proteins with diverse functions (30), thereby suggesting that PAM2

230

participated in protein–protein interactions in a wide range of cellular processes. Two of

231

the three PxxY motifs were typical binding regions of the WW domains of the Nedd4

232

family members (31) and were located in the PAM2 motif of Paip1 (Fig. 1B). Paip1

233

interacted with PABP via the PAM2 motif. We measured the interaction of Paip1 and

234

PABP upon WWP2 silencing and overexpression to determine whether or not WWP2 and

235

PABP competed for Paip1 binding. WWP2 did not affect the interaction of Paip1 and

236

PABP (Fig. 3F), thereby suggesting that Paip1’s PAM2 motif (comprising amino acids

237

116 to 143) sufficiently interacted with the WW domain of WWP2.

238

Degradation and ubiquitination of Paip1 by WWP2 depends on the presence of

239

the PEFYPSGY sequence. Based on data from previous studies, we speculated that the

240

two PxxY motifs (PEFYPSGY sequence) in PAM2 participated in WWP2’s recognition

241

and degradation of Paip1. We transfected different Paip1 deletion mutants into HEK293T

242

cells with Flag-WWP2-WT to determine the importance of the two PxxY motifs.

243

Degradation analysis showed that mutants 144 to 479 included one PxxY motif that was

244

not degraded by overexpressed WWP2. By contrast, the N-terminal mutants 1 to 143,

245

which comprised the first two PxxY motifs, were efficiently degraded by WWP2 (Fig.

246

4A). This phenomenon suggested that the critical degradation signal was present in the 12

247

mutants. To further determine which PxxY motif was critical for WWP2-mediated

248

degradation, we used the ΔPEFY, ΔPSGY, and ΔPEFYPSGY forms of Paip1 to analyze

249

protein levels in the presence or absence of WWP2. Paip1-ΔPEFY and Paip1-ΔPSGY

250

were both downregulated by WWP2. However, WWP2 did not downregulate

251

Paip1-ΔPEFYPSGY (Fig. 4B). The two PxxY motifs and the PEFYPSGY sequence were

252

necessary for WWP2 to recognize Paip1.

253

We performed an ubiquitination assay to test whether or not the resistance of the

254

mutant Paip1-ÄPEFYPSGY to WWP2-mediated degradation was caused by abrogated

255

ubiquitination. WWP2 did not ubiquitylate the truncate (Fig. 4C). The PEFYPSGY

256

sequence was critical in the catalytic activity of WWP2 during Paip1 ubiquitination and

257

degradation.

258

WWP2 negatively regulates Paip1-mediated translation enhancement. To gain

259

insight into the functional relationship between WWP2 and Paip1, we initially determined

260

the translational stimulatory activity via in vivo translation assays. Under the control of

261

the Tet-off promoter expressing WWP2-WT or WWP2-C838 A, DNA vectors were

262

transfected into HeLa cells along with constructs expressing Renilla luciferase and tTA.

263

Each construct yielded comparable amounts of protein. Paip1 was absent in the extract

264

expressing WWP2 (Fig. 5A). The relative induction of luciferase activity was determined

265

by calculating the ratio of Renilla luciferase activity between induced (without Tet) and

266

repressed (with Tet at 300 ng/ml) expressions of WWP2. Luciferase activity was lower

267

with the overexpression of the wild-type WWP2 plasmid than with the expression of the 13

268

vector alone (Fig. 5B lanes 1, 2, 3, 4, and 5). However, the overexpression of

269

WWP2-C838 A did not affect in vivo translation (Fig. 5B lanes 6 and 7), thereby

270

suggesting that the ligase activity of WWP2 was required. WWP2 overexpression did not

271

affect Renilla luciferase mRNA levels (Fig. 5C).

272

To determine whether or not the translational suppression induced by WWP2

273

depended on the presence of Paip1, we examined the effect of Paip1 silencing via siRNA

274

on WWP2 activity. The expressions of WWP2 and Paip1 were confirmed by Western

275

blot analysis (Fig. 5D). No significant translational repression was observed when Paip1

276

was knocked down compared with the control (Fig. 5E). Renilla luciferase mRNA levels

277

were not affected by siRNA treatments (Fig. 5F). Therefore, WWP2 repressed mRNA

278

translation depending on the presence of Paip1. These data confirmed the importance of

279

the protein level regulation of Paip1 by WWP2. We demonstrated that WWP2 is a novel

280

regulator of translational initiation.

281 282

Discussion

283

Translation is important in the regulation of gene expression and is implicated in the

284

control of cell growth, proliferation, and differentiation (32–34). In eukaryotes, initiation

285

is the rate-limiting step of translation under most circumstances; initiation is a major

286

target for regulation (33). Paip1 is a mammalian PABP that binds to eIF4A and eIF3 and

287

stimulates translational initiation. In the present study, we showed that Paip1 protein was

288

degraded by HECT ubiquitin ligase WWP2. The following findings from the present 14

289

study directly corroborate the use of Paip1 as a physiological substrate of WWP2. WWP2

290

directly interacted with Paip1, and the interaction depended on the integrity of the WW

291

domain of WWP2. The loss of WWP2 impeded Paip1 turnover. WWP2 promoted Paip1

292

ubiquitination both in vivo and in a reconstituted in vitro system. The ubiquitin ligase

293

activity of WWP2 and the PxxY motif of Paip1 were critical for ubiquitination and

294

degradation. Previous studies have demonstrated the involvement of WWP2 in the

295

regulation of transcription, embryonic stem cell development, cellular transport, T-cell

296

activation processes, and tumorigenesis by targeting distinct substrates (35). In the

297

present study, we revealed that Paip1 was a novel substrate of WWP2. We also

298

emphasized the importance of WWP2 in the regulation of translation.

299

The involvement of deregulation of translational initiation in cancer development and

300

progression became the focus of various studies only recently. WWP2 mediates the

301

depletion of phosphatase and tensin homolog and consequently elevates

302

phosphoinositide-3-kinase–protein kinase B pathway activity (36). Such regulatory

303

activities may enhance eIF4E function and activate cap-dependent translation initiation,

304

leading to the selective increase of translation of key mRNAs; these mRNAs are involved

305

in tumor growth, angiogenesis, and cell survival (37). Thus, we hypothesized that

306

WWP2-mediated degradation of Paip1 selectively reduced the expression of numerous

307

potent growth and survival factors. The role of WWP2 in tumorigenesis, which depends

308

on WWP2’s regulation of translation, requires further investigation.

309

Circularization of the mRNA by bridging the mRNA 5’ and 3’ is an important step in 15

310

the process of translation initiation. In yeast, eIF4G interacts with PABP to contribute to

311

the circularization of the mRNA and mediation of the poly(A) tail-dependent translation

312

(11). In plants, PABP binds to eIF-iso4G and eIF4B, thereby increasing the RNA-binding

313

activity of PABP (13). However, mammalian cells possess dual systems, namely,

314

PABP–Paip1 and PABP–eIF4G. These systems regulate the formation of mRNP. The

315

presence of Paip1 in animal cells reflects evolutionary advancement and allows higher

316

eukaryotes to link PABP to eIF4A function. Information is lacking on whether the

317

degradation of Paip1 by WWP2 reduces the rate of the circular mRNA conformation and

318

inhibits mRNA translation. Paip1’s sequence and function are similar to those of the

319

eIF4G protein. WWP2 may also regulate the protein level of eIF4G in human cells, but

320

the relationship between WWP2 and eIF4G requires further study.

321

The stimulation of translation by Paip1 in vivo decreased upon the deletion of the

322

N-terminal sequence containing PABP and the eIF3 binding domain known as PAM2

323

(22). In the present study, PAM2 was critical for the binding of PABP to WWP2.

324

Paip1-interacting proteins can potentially compete for Paip1 binding via the PAM2

325

domain, thereby regulating translation. Further studies are required to determine the

326

presence of an overlap among the binding sites of WWP2, eIF3, and PABP in Paip1.

327

WWP2 was identified as a typical ubiquitin ligase for Paip1. We showed that the

328

overexpression of WWP2 significantly decreased Paip1 protein level, and such decrease

329

suppressed translation. We also discovered that the WW domain of WWP2 interacted

330

with Paip1 fragments, which contained two PxxY motifs. The degradation and 16

331

ubiquitination of Paip1 by WWP2 was transiently abolished by the deletion of the two

332

PxxY motifs. WWP2 is an important translational suppressor that participates in Paip1

333

degradation. The function of WWP2 in the regulation of translation was clarified for the

334

first time. The mechanisms underlying WWP2’s regulation of Paip1 should be studied

335

further to reveal the details of WWP2’s function.

336 337

ACKNOWLEDGMENTS

338

This work was supported by grants from the Nature Science of Shandong Province,

339

Q2008C08. The funders had no role in the study design, data collection, data analysis,

340

decision to publish, or preparation of the manuscript.

341 342

REFERENCES

343 344 345 346 347 348 349 350 351 352 353 354 355 356 357 358 359

1.

Parsyan A, Hernandez G, Meterissian S. 2012. Translation initiation in colorectal cancer. Cancer Metastasis Rev 31:387-395.

2.

Jia Y, Polunovsky V, Bitterman PB, Wagner CR. 2012. Cap-dependent translation initiation factor eIF4E: an emerging anticancer drug target. Med Res Rev 32:786-814.

3.

Graff JR, Konicek BW, Carter JH, Marcusson EG. 2008. Targeting the eukaryotic translation initiation factor 4E for cancer therapy. Cancer research 68:631-634.

4.

Cheung YN, Maag D, Mitchell SF, Fekete CA, Algire MA, Takacs JE, Shirokikh N, Pestova T, Lorsch JR, Hinnebusch AG. 2007. Dissociation of eIF1 from the 40S ribosomal subunit is a key step in start codon selection in vivo. Genes Dev 21:1217-1230.

5.

Chiu WL, Wagner S, Herrmannova A, Burela L, Zhang F, Saini AK, Valasek L, Hinnebusch AG. 2010. The C-terminal region of eukaryotic translation initiation factor 3a (eIF3a) promotes mRNA recruitment, scanning, and, together with eIF3j and the eIF3b RNA recognition motif, selection of AUG start codons. Molecular and cellular biology 30:4415-4434.

6.

Pisarev AV, Unbehaun A, Hellen CU, Pestova TV. 2007. Assembly and analysis of eukaryotic translation initiation complexes. Methods Enzymol 430:147-177.

7.

Lorsch JR, Dever TE. 2010. Molecular view of 43 S complex formation and start site selection in eukaryotic translation initiation. THE JOURNAL OF BIOLOGICAL CHEMISTRY 285:21203-21207. 17

360 361 362 363 364 365 366 367 368 369 370 371 372 373 374 375 376 377 378 379 380 381 382 383 384 385 386 387 388 389 390 391 392 393 394 395 396 397 398 399 400

8.

Preiss T, M WH. 2003. Starting the protein synthesis machine: eukaryotic translation initiation. Bioessays 25:1201-1211.

9.

Prevot D, Darlix JL, Ohlmann T. 2003. Conducting the initiation of protein synthesis: the role of eIF4G. Biol Cell 95:141-156.

10.

Kessler SH, Sachs AB. 1998. RNA recognition motif 2 of yeast Pab1p is required for its functional interaction with eukaryotic translation initiation factor 4G. Molecular and cellular biology 18:51-57.

11.

Tarun SZ, Jr., Sachs AB. 1996. Association of the yeast poly(A) tail binding protein with translation initiation factor eIF-4G. The EMBO Journal 15:7168-7177.

12.

Tarun SZ, Jr., Wells SE, Deardorff JA, Sachs AB. 1997. Translation initiation factor eIF4G mediates in vitro poly(A) tail-dependent translation. Proceedings of the National Academy of Sciences of the United States of America 94:9046-9051.

13.

Le H, Tanguay RL, Balasta ML, Wei CC, Browning KS, Metz AM, Goss DJ, Gallie DR. 1997. Translation initiation factors eIF-iso4G and eIF-4B interact with the poly(A)-binding protein and increase its RNA binding activity. THE JOURNAL OF BIOLOGICAL CHEMISTRY 272:16247-16255.

14.

Khaleghpour K, Kahvejian A, De Crescenzo G, Roy G, Svitkin YV, Imataka H, O'Connor-McCourt M, Sonenberg N. 2001. Dual interactions of the translational repressor Paip2 with poly(A) binding protein. Molecular and cellular biology 21:5200-5213.

15.

Roy G, De Crescenzo G, Khaleghpour K, Kahvejian A, O'Connor-McCourt M, Sonenberg N. 2002. Paip1 interacts with poly(A) binding protein through two independent binding motifs. Mol Cell Biol 22:3769-3782.

16.

Imataka H, Olsen HS, Sonenberg N. 1997. A new translational regulator with homology to eukaryotic translation initiation factor 4G. The EMBO Journal 16:817-825.

17.

Levy-Strumpf N, Deiss LP, Berissi H, Kimchi A. 1997. DAP-5, a novel homolog of eukaryotic translation initiation factor 4G isolated as a putative modulator of gamma interferon-induced programmed cell death. Molecular and cellular biology 17:1615-1625.

18.

Yamanaka S, Poksay KS, Arnold KS, Innerarity TL. 1997. A novel translational repressor mRNA is edited extensively in livers containing tumors caused by the transgene expression of the apoB mRNA-editing enzyme. Genes Dev 11:321-333.

19.

Morino S, Imataka H, Svitkin YV, Pestova TV, Sonenberg N. 2000. Eukaryotic translation initiation factor 4E (eIF4E) binding site and the middle one-third of eIF4GI constitute the core domain for cap-dependent translation, and the C-terminal one-third functions as a modulatory region. Molecular and cellular biology 20:468-477.

20.

Imataka H, Sonenberg N. 1997. Human eukaryotic translation initiation factor 4G (eIF4G) possesses two separate and independent binding sites for eIF4A. Molecular and cellular biology 17:6940-6947.

21.

Craig AW, Haghighat A, Yu AT, Sonenberg N. 1998. Interaction of polyadenylate-binding protein with the eIF4G homologue PAIP enhances translation. Nature 392:520-523.

22.

Martineau Y, Derry MC, Wang X, Yanagiya A, Berlanga JJ, Shyu AB, Imataka H, Gehring K, Sonenberg N. 2008. Poly(A)-binding protein-interacting protein 1 binds to eukaryotic translation initiation factor 3 to stimulate translation. Mol Cell Biol 28:6658-6667. 18

401 402 403 404 405 406 407 408 409 410 411 412 413 414 415 416 417 418 419 420 421 422 423 424 425 426 427 428 429 430 431 432 433 434 435 436 437

23.

Martineau Y, Wang X, Alain T, Petroulakis E, Shahbazian D, Fabre B, Bousquet-Dubouch MP, Monsarrat B, Pyronnet S, Sonenberg N. 2014. Control of Paip1-eukayrotic translation initiation factor 3 interaction by amino acids through S6 kinase. Mol Cell Biol 34:1046-1053.

24.

Zou W, Chen X, Shim JH, Huang Z, Brady N, Hu D, Drapp R, Sigrist K, Glimcher LH, Jones D. 2011. The E3 ubiquitin ligase Wwp2 regulates craniofacial development through mono-ubiquitylation of Goosecoid. Nature Cell Biology 13:59-65.

25.

Nakamura Y, Yamamoto K, He X, Otsuki B, Kim Y, Murao H, Soeda T, Tsumaki N, Deng JM, Zhang Z, Behringer RR, Crombrugghe B, Postlethwait JH, Warman ML, Nakamura T, Akiyama H. 2011. Wwp2 is essential for palatogenesis mediated by the interaction between Sox9 and mediator subunit 25. Nature Communications 2:251.

26.

Maddika S, Kavela S, Rani N, Palicharla VR, Pokorny JL, Sarkaria JN, Chen J. 2011. WWP2 is an E3 ubiquitin ligase for PTEN. Nature Cell Biology 13:728-733.

27.

Soond SM, Chantry A. 2011. Selective targeting of activating and inhibitory Smads by distinct WWP2 ubiquitin ligase isoforms differentially modulates TGFbeta signalling and EMT. Oncogene 30:2451-2462.

28.

Gossen M, Bujard H. 1992. Tight control of gene expression in mammalian cells by tetracycline-responsive promoters. Proceedings of the National Academy of Sciences of the United States of America 89:5547-5551.

29.

Bernassola F, Karin M, Ciechanover A, Melino G. 2008. The HECT family of E3 ubiquitin ligases: multiple players in cancer development. Cancer Cell 14:10-21.

30.

Albrecht M, Lengauer T. 2004. Survey on the PABC recognition motif PAM2. Biochem Biophys Res Commun 316:129-138.

31.

Ingham RJ, Gish G, Pawson T. 2004. The Nedd4 family of E3 ubiquitin ligases: functional diversity within a common modular architecture. Oncogene 23:1972-1984.

32. 33.

Conlon I, Raff M. 1999. Size control in animal development. Cell 96:235-244. Gingras AC, Raught B, Sonenberg N. 1999. eIF4 initiation factors: effectors of mRNA recruitment to ribosomes and regulators of translation. Annu Rev Biochem 68:913-963.

34.

Miron M, Verdu J, Lachance PE, Birnbaum MJ, Lasko PF, Sonenberg N. 2001. The translational inhibitor 4E-BP is an effector of PI(3)K/Akt signalling and cell growth in Drosophila. Nature Cell Biology 3:596-601.

35.

Chen A, Gao B, Zhang J, McEwen T, Ye SQ, Zhang D, Fang D. 2009. The HECT-type E3 ubiquitin ligase AIP2 inhibits activation-induced T-cell death by catalyzing EGR2 ubiquitination. Molecular and cellular biology 29:5348-5356.

36.

Chantry A. 2011. WWP2 ubiquitin ligase and its isoforms: new biological insight and promising disease targets. Cell cycle 10:2437-2439.

37.

Graff JR, Konicek BW, Carter JH, Marcusson EG. 2008. Targeting the eukaryotic translation initiation factor 4E for cancer therapy. Cancer Res 68:631-634.

438 439 19

440

FIGURE LEGENDS

441

Fig. 1. WWP2 negatively regulates Paip1 protein level. (A) MG132 treatment upregulates

442

Paip1 protein level. HEK293T cells were treated with MG132 (20 µM) for 8 h before

443

harvest. Endogenous Paip1, PABP, and Paip2 levels were analyzed by immunoblotting.

444

GAPDH was used as an internal control. (B) Schematic of Paip1 protein. The positions of

445

PxxY motifs are labeled. (C) WWP2 specifically reduces Paip1 protein amount. The

446

indicated Nedd4 family of E3 ligases was co-transfected with Paip1 into HEK293T cells,

447

and cell lysates were analyzed by immunoblotting. (D) WWP2 downregulates Paip1 in a

448

proteasome-dependent manner. Increasing concentrations of Flag-WWP2 plasmids were

449

co-transfected into HEK293T cells with Myc-Paip1. Cells were treated with MG132

450

(20 μM) and lactacystin (30 μM) or dimethyl sulfoxide (DMSO) for 16 h. The cell lysates

451

were analyzed by immunoblotting. (E) WWP2 decreases the Paip1 protein level in a

452

dose-dependent manner. Cells were transfected with increasing amounts of WWP2-WT

453

or the catalytic mutant form WWP2-C838A. Subsequently, Paip1 level was detected. (F)

454

WWP2 can not affect the mRNA level of Paip1. Paip1 mRNA prepared from the

455

transfected HEK293T cells was analyzed by real-time PCR assay. Data are presented as

456

mean ± S.D. (n = 3). (G) WWP2 depletion increases the endogenous Paip1 protein level.

457

HEK293T and HeLa cells were transfected with non-targeted control or with

458

WWP2-specific siRNA. The endogenous WWP2 and Paip1 levels were analyzed by

459

Western blot. (H) WWP2 depletion can not affect the Paip1 mRNA level. Paip1 mRNA

460

prepared from the transfected HEK293T and HeLa cells was analyzed by real-time PCR 20

461

assay. Data are presented as mean ± S. D. (n = 3). (I) WWP2 can not affect the

462

endogenous Paip2 and PABP protein levels. Cells were transfected with the WWP2-WT

463

or the catalytic mutant form WWP2-C838A. Paip2 and PABP protein levels were

464

detected by immunoblotting (top). HEK293T cells were transfected with non-targeted

465

control or WWP2–specific siRNA. The endogenous WWP2 and PABP protein levels

466

were analyzed by immunoblotting (bottom). (J) Overexpression of Paip2 can not affect

467

WWP2-mediated degradation of Paip1. HEK293T cells were transfected with the vector

468

or with Paip2. At 24 h after transfection, cells were transfected with WWP2 and analyzed

469

by immunoblotting.

470

Fig. 2. WWP2 promotes the ubiquitination and degradation of Paip1. (A) WWP2 reduces

471

the half-life of endogenous Paip1 protein. HEK293T cells were transfected with control

472

plasmid or Flag-WWP2-WT or the catalytic mutant form Flag-WWP2-C838 A, and cells

473

were treated with the protein synthesis inhibitor CHX (10 µg/ml) for the indicated

474

durations before harvest. Paip1 protein expression was analyzed. Quantitative analysis

475

was performed by measuring integrated optical density using the program Gel-Pro

476

analyzer. Data are presented as mean ± S. D. (n = 3). (B) Depletion of WWP2 prolonges

477

the half-life of endogenous Paip1 protein. HEK293T cells were transfected with control

478

siRNA or WWP2 siRNA and treated with CHX. Paip1 protein expression was

479

subsequently analyzed. Quantitative analysis was performed by measuring integrated

480

optical density using the program Gel-Pro analyzer. Data are presented as mean ± S. D. (n

481

= 3). (C) WWP2 catalyzes the ubiquitination of Paip1 in vitro. A mixture comprising 21

482

purified HA-ubiquitin, E1, E2 (UbcH5c), bacteria-expressed and purified

483

His-WWP2-WT or C838A, and GST-Paip1 or GST was used for in vitro ubiquitination

484

assays and subsequent immunoblotting with anti-HA. (D) WWP2 enhances the

485

ubiquitination of Paip1 in vivo. HEK293T cells were transfected with HA-Ub,

486

Myc-Paip1, control vector, or Flag-WWP2, and treated with MG132 as indicated.

487

Ubiquitinated Paip1 was immunoprecipitated by anti-Myc antibody and analyzed by

488

immunoblotting. (E) WW and HECT domains of WWP2 were involved in the

489

ubiquitination of Paip1. HA-Ub, Myc-Paip1, and WWP2 constructs were co-transfected

490

into HEK293T cells and treated with MG132. Ubiquitinated Paip1 was

491

immunoprecipitated with anti-HA antibody and analyzed by immunoblotting.

492

Fig. 3. Mapping of the interaction region between the truncated domains of WWP2 and

493

Paip1. (A) WWP2 was colocalized with Paip1 within the cytoplasm. Myc-Paip1 and

494

Flag-WWP2 were co-transfected into HEK293T cells, and cells were stained 24 h later

495

using mouse anti-Flag and rabbit anti-Myc antibodies for visualization by confocal

496

microscopy. (B) Paip1 was co-immunoprecipitated with wild type and catalytic mutant

497

forms of WWP2. Wild type and catalytic mutant forms of WWP2 and Myc-Paip1 were

498

transfected into HEK293T cells. Cell lysates were immunoprecipitated with anti-Flag

499

antibody. Lysates and immunoprecipitates were analyzed by immunoblotting. (C)

500

Immunoprecipitation analysis of WWP2 and Paip1 in vivo. HeLa cells were transfected

501

with negative control siRNA or siRNA targeted Paip1, and cells were harvested after 36 h.

502

The extracts were immunoprecipitated with anti-WWP2 antibody and subsequently 22

503

analyzed by immunoblotting using anti-WWP2, anti-Paip1, anti-eIF3a, and anti-PABP

504

antibodies. Pre-immune IgG was used as the control. (D) Direct interaction between the

505

WW domain of WWP2 and Paip1. GST pull-downs were conducted with GST or

506

GST-WWP2 fragments along with purified His-Paip1. Input and pull-down samples were

507

subjected to immunoblotting with anti-GST and anti-His antibodies. Input represented 10%

508

of the sample used for the pull-down. (E) PAM2 motif of Paip1 showed direct binding to

509

WWP2. GST pull-downs were performed with GST or GST-Paip1 fragments along with

510

purified His-WWP2. Input and pull-down samples were subjected to immunoblotting

511

with anti-GST and anti-His antibodies. Input represented 10% of the sample used for

512

pull-down. (F) WWP2 can not affect the interaction between Paip1 and PABP. HEK293T

513

cells were transfected with Flag-WWP2-WT or the catalytic mutant form

514

Flag-WWP2-C838A. Cell lysates were immunoprecipitated with anti-Paip1 antibody.

515

Both the lysate and immunoprecipitates were analyzed by immunoblotting (left).

516

HEK293T cells were transfected with control siRNA or WWP2 siRNA. Cell lysates were

517

immunoprecipitated with anti-Paip1 antibody. Lysates and immunoprecipitates were

518

analyzed by immunoblotting (right).

519

Fig. 4. PEFYPSGY sequence is sufficient for the ubiquitination and degradation of Paip1.

520

(A) HEK293T cells were transfected with plasmids expressing either full-length Paip1 or

521

mutants of Paip1 with or without a plasmid encoding Flag-WWP2-WT. (B) PEFYPSGY

522

sequence maintains the WWP2-mediated degradation of Paip1. HEK293T cells were

523

transfected with plasmids expressing Paip1-ΔPEFY, Paip1-ΔPSGY, or 23

524

Paip1-ΔPEFYPSGY with or without Flag-WWP2. The cell lysates were analyzed by

525

immunoblotting. (C) PEFYPSGY sequence attenuated WWP2-mediated ubiquitination of

526

Paip1. HEK293T cells were co-transfected with Myc-Paip1 deletions and HA-Ub with or

527

without Flag-WWP2. Ubiquitinated Paip1 was immunoprecipitated with an anti-Myc

528

antibody and protein A/G-agarose beads under denaturing conditions to eliminate any

529

WWP2-associated protein by non-covalent bonding.

530

Fig. 5. Overexpression of WWP2 represses translation via the Paip1 protein. (A) Cells

531

were transfected with the indicated pTet-HA-WWP2-WT or catalytic mutant form

532

pTet-HA-WWP2-C838A plasmids along with constructs expressing Renilla luciferase

533

and tTA. Cells were cultured in a medium containing 0 or 300 ng/ml of Tet. Cells were

534

harvested and subjected to Western blot analysis with the indicated antibodies. (B)

535

Renilla luciferase activity was quantified in HeLa cell extracts harvested in (A) and

536

normalized on the total protein level. Relative induction of the luciferase reporter was

537

determined by calculating the ratio of Renilla luciferase activity between induced

538

(without Tet) and repressed (with 300 ng/ml Tet) expression of the indicated HA-WWP2.

539

Error bars denote the standard error of the mean for the three independent experiments.

540

(C) Quantitative real-time PCR of Renilla luciferase mRNA obtained from total RNA of

541

duplicate HeLa cells harvested in (A). Data are presented as mean ± S. D. (n = 3). (D)

542

HeLa cells were transfected with the indicated siRNA sequences. At 24 h after siRNA

543

transfection, cells were transfected as described in (A). Cells were subsequently placed in

544

a medium containing 0 or 300 ng/ml of Tet. Cells were harvested and subjected to 24

545

Western blot analysis with the indicated antibodies. (E) Renilla luciferase activity was

546

quantified in HeLa cell extracts harvested in (D) and normalized on the total protein level.

547

Relative induction of the luciferase reporter was determined by calculating the ratio of

548

Renilla luciferase activity between induced (without Tet) and repressed (with 300 ng/ml

549

Tet) expression of the indicated HA-WWP2. Error bars denote the standard error of the

550

mean for three independent experiments. (F) Quantitative RT–PCR of Renilla luciferase

551

mRNA obtained from the total RNA of the duplicate HeLa cells harvested in (D). Data

552

are presented as mean ± S. D. (n = 3).

25

Paip1, an effective stimulator of translation initiation, is targeted by WWP2 for ubiquitination and degradation.

Poly(A)-binding protein-interacting protein 1 (Paip1) stimulates translational initiation by inducing the circularization of mRNA. However, the mechan...
984KB Sizes 0 Downloads 5 Views